SOX1 Promotes Differentiation of Nasopharyngeal Carcinoma Cells by Activating Retinoid Metabolic Pathway

Total Page:16

File Type:pdf, Size:1020Kb

SOX1 Promotes Differentiation of Nasopharyngeal Carcinoma Cells by Activating Retinoid Metabolic Pathway Lei et al. Cell Death and Disease (2020) 11:331 https://doi.org/10.1038/s41419-020-2513-1 Cell Death & Disease ARTICLE Open Access SOX1 promotes differentiation of nasopharyngeal carcinoma cells by activating retinoid metabolic pathway Xin-Xing Lei1,YunLiu2, Jin-Xing Wang3,QianCai2,MinYan1,Hui-PingHe1,QuentinLiu1,3,Zi-JieLong3 and Zhong Guan2 Abstract Undifferentiation is a key feature of nasopharyngeal carcinoma (NPC), which presents as a unique opportunity for intervention by differentiation therapy. In this study, we found that SOX1 inhibited proliferation, promoted differentiation, and induced senescence of NPC cells, which depended on its transcriptional function. RNA-Seq- profiling analysis showed that multiple undifferentiated markers of keratin family, including KRT5, KRT13, and KRT19, were reduced in SOX1 overexpressed NPC cells. Interestingly, gene ontology (GO) analysis revealed genes in SOX1 overexpressed cells were enriched in extracellular functions. The data of LC/MS untargeted metabolomics showed that the content of retinoids in SOX1 overexpressed cells and culture medium was both higher than that in the control group. Subsequently, we screened mRNA level of genes in retinoic acid (RA) signaling or metabolic pathway and found that the expression of UDP-glucuronosyltransferases was significantly decreased. Furtherly, UGT2B7 could rescue the differentiation induced by SOX1 overexpression. Inhibition of UGTs by demethylzeylasteral (T-96) could mimic SOX1 to promote the differentiation of NPC cells. Thus, we described a mechanism by which SOX1 regulated 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; the differentiation of NPC cells by activating retinoid metabolic pathway, providing a potential target for differentiation therapy of NPC. Introduction offer new opportunities for treating NPC. Recently, Nasopharyngeal carcinoma (NPC) is endemic in remarkable achievements have been made by differ- South-Eastern Asia, and northern Africa. At present, the entiation therapy in the treatment of some types of main treatment approaches for NPC include radio- tumors3,4. 95% of NPC is pathologically undifferentiated therapy, chemotherapy, and targeted therapy. Radiation squamous cell carcinoma, which make NPC an appro- therapy is the firstchoiceforthetreatmentofNPC. priate model for differentiation therapy. However, it is inevitable that nasopharyngeal mucosa SOX1 encodes a member of the SOX (SRY-related and tissue will be damaged after radiotherapy1,2. Thus, high mobility group box) family of transcription factors more effective and less toxic regimens are needed to involved in the regulation of embryonic development – and determination of the cell fate5 7.SOX1shares similar biochemical properties and functions with SOX2 Correspondence: Quentin Liu ([email protected])or or other members, but the different affinity to target Zi-Jie Long ([email protected]) or Zhong Guan ([email protected]) genes, post-transcriptional modifications and interac- 1Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, 510060 Guangzhou, China tions with other co-factors lead to different functions in – 2Department of Otorhinolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat- distinct biological contexts8 10. SOX proteins bind sen University, 510120 Guangzhou, China sequence-specifically to DNA by a high-mobility group Full list of author information is available at the end of the article These authors contributed equally: Xin-Xing Lei, Yun Liu (HMG) domain that allows them to function as Edited by A. Stephanou © The Author(s) 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/. Official journal of the Cell Death Differentiation Association Lei et al. Cell Death and Disease (2020) 11:331 Page 2 of 15 transcription factors8,11. Gene silencing in cancer is V5-tagged SOX1, ΔHMG SOX1, or Δ246-391 SOX1. The associated with promoter hypermethylation12 and SOX1 primers used for gene cloning and plasmid construction hypermethylation was detected in primary tumor tis- are listed in Supplementary Table S1. sues13. SOX1 inhibits cell proliferation, reduces migra- tion and invasion ability, and induces differentiation of Lentiviral production, infections, and cell line generation – tumor cells14 17. Recently, we found that SOX1 was Lentivirus was produced by transient transfection by associated with decreased expression of undifferentiation Lipofectamine 2000 (Invitrogen) in human embryonic markers (KRT13 and KRT19) in NPC cells16. However, kidney (HEK) 293T cells using with a second-generation the mechanism by which SOX1 promotes NPC cell dif- lentiviral vector system. All virus-containing medium was ferentiation has not been established. mixed with 8 μg/mL polybrene (Sigma-Aldrich). Virus Retinoic acid (RA), especially all-trans-retinoic acid produced from pLVX-Tet3G plasmid was used to infect (ATRA), is the most potent natural metabolite of vitamin wild type HONE1 or CNE2 cells to construct HONE1- A. RA is involved in a variety of biological functions Tet-On or CNE2-Tet-On stable cell lines. Cells were including embryogenesis, cell differentiation, and apop- selected in 1 mg/mL G418 for at least 2 weeks. Subse- tosis18,19. Retinoids induce differentiation and/or apop- quently, HONE1-Tet-On and CNE2-Tet-On cell lines tosis in tumor cells and show anti-proliferative and anti- were infected by virus produced from pLVX-TRE-(X) oxidant activity, which have great potential as therapeutic plasmids and selected by 2 μg/mL puromycin for 6 days. or preventive agents. The distribution and level of RA in HONE1TRE-SOX1 and CNE2TRE-SOX1 cells infected by virus embryonic tissues are tightly controlled by oxidation produced from pLVX-UGT1A6 or pLVX-UGT2B7 plas- through retinol and retinal dehydrogenases and by mids were selected by 12 μg/mL blasticdin for 7 days. hydroxylation via specific cytochrome P450 family 26 enzymes (CYP26s)20,21. Cell culture and doxycycline induction UDP-glucuronosyltransferases (UGTs) are phase II The HONE1 and CNE2 cell lines were obtained from metabolism isoenzymes that are found to be active in Dr.Chao-NanQian(SunYat-senUniversity,Guangz- liver, kidneys, epithelial cells of the lower gastrointestinal hou, China). Wild type cell lines and their lentiviral- tract and brain20,22. Although the characterizations of infected stable cell lines were all maintained in RPMI UGT gene family members were identified, their roles in 1640 (Invitrogen) supplemented with 10% fetal bovine clearance and homeostasis of endogenous substrates are serum (FBS, Hyclone). The cells were incubated at 37 °C still insufficiently understood. Previous study proves that in a humidified chamber containing 5% CO2.HONE1- retinoids and their analogs are glucuronidated by Tet-On-(X) and CNE2-Tet-On-(X) cell lines were trea- UGTs20,23,24. Excess retinoids can also inhibit the ted with 2 μg/mL doxycycline (Medchem Express) to expression of UGTs, indicating the existence of regulatory induce overexpression of SOX1 or its mutant/ loops and complexity25. truncated form. In the present study, we showed that overexpression of SOX1 promoted NPC cells to differentiate via its tran- Cell viability assay scriptional function. These new data also established a Cells were seeded onto 96-well plates at an initial mechanism that retinoids were accumulated by SOX1, density of 2 × 103 cells/well. At specified time points, contributing to the differentiation of NPC cells, which 10 μL of CCK-8 solution was added to each well of the offered a novel approach for NPC differentiation therapy. plate. Then the plate was incubated for another 2 h. Cell viability was determined by scanning with a microplate Materials and methods reader at 450 nm. Plasmid constructs The plasmids encoding human SOX1 (wild type, WT), Immunofluorescence staining mutant SOX1 (SOX1mut_2), V5-tagged SOX1, ΔHMG Cells were fixed in 4% paraformaldehyde at room SOX1, and Δ246-391 SOX1 were generated by PCR temperature for 10 min and permeabilized in 0.5% Tri- amplification and subcloned into the pLVX-TRE3G ton X-100 in PBS for 10 min. Slides were incubated with expression vector; UGT1A6 and UGT2B7 were sub- the primary antibody (1:200 dilution) overnight. Immune cloned into pLVX expression vector; promoters of complexes were stained with the secondary antibody UGT1A6 and UGT2B7 were subcloned into pGL3-basic conjugated to Alexa-488 or Alexa-546 (Invitrogen, 1:200 vector using the ClonExpress II One Step Cloning Kit or dilution) at room temperature for 1 h. Nuclei were ClonExpress MultiS One Step Cloning Kit (Vazyme) stained with DAPI (Sigma-Aldrich) and viewed with an according to the manufacturer’s instructions. The new Olympus IX71 microscope. The following antibodies plasmid was named
Recommended publications
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [Show full text]
  • Ncomms2254.Pdf
    ARTICLE Received 3 May 2012 | Accepted 5 Nov 2012 | Published 4 Dec 2012 DOI: 10.1038/ncomms2254 The RB family is required for the self-renewal and survival of human embryonic stem cells Jamie F. Conklin1,2,3, Julie Baker2,3 & Julien Sage1,2,3 The mechanisms ensuring the long-term self-renewal of human embryonic stem cells are still only partly understood, limiting their use in cellular therapies. Here we found that increased activity of the RB cell cycle inhibitor in human embryonic stem cells induces cell cycle arrest, differentiation and cell death. Conversely, inactivation of the entire RB family (RB, p107 and p130) in human embryonic stem cells triggers G2/M arrest and cell death through functional activation of the p53 pathway and the cell cycle inhibitor p21. Differences in E2F target gene activation upon loss of RB family function between human embryonic stem cells, mouse embryonic stem cells and human fibroblasts underscore key differences in the cell cycle regulatory networks of human embryonic stem cells. Finally, loss of RB family function pro- motes genomic instability in both human and mouse embryonic stem cells, uncoupling cell cycle defects from chromosomal instability. These experiments indicate that a homeostatic level of RB activity is essential for the self-renewal and the survival of human embryonic stem cells. 1 Department of Pediatrics, Stanford Medical School, Stanford, California 94305, USA. 2 Department of Genetics, Stanford Medical School, Stanford, California 94305, USA. 3 Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medical School, Stanford, California 94305, USA. Correspondence and requests for materials should be addressed to J.S.
    [Show full text]
  • Author Manuscript Faculty of Biology and Medicine Publication
    Serveur Académique Lausannois SERVAL serval.unil.ch Author Manuscript Faculty of Biology and Medicine Publication This paper has been peer-reviewed but does not include the final publisher proof-corrections or journal pagination. Published in final edited form as: Title: Sexual dimorphism in cancer. Authors: Clocchiatti A, Cora E, Zhang Y, Dotto GP Journal: Nature reviews. Cancer Year: 2016 May Volume: 16 Issue: 5 Pages: 330-9 DOI: 10.1038/nrc.2016.30 In the absence of a copyright statement, users should assume that standard copyright protection applies, unless the article contains an explicit statement to the contrary. In case of doubt, contact the journal publisher to verify the copyright status of an article. Sexual dimorphism in cancer Andrea Clocchiatti1+, Elisa Cora2+, Yosra Zhang1,2 and G. Paolo Dotto1,2* 1Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; 2Department of Biochemistry, University of Lausanne, Epalinges, CH-1066, Switzerland + These authors contributed equally to the work * Corresponding author: G. Paolo Dotto email: [email protected] 1 Abstract The incidence of many cancer types is significantly higher in the male than female populations, with associated differences in survival. Occupational and/or behavioral factors are well known underlying determinants. However, cellular/molecular differences between the two sexes are also likely to be important. We are focusing here on the complex interplay that sexual hormones and sex chromosomes can have in intrinsic control of cancer initiating cell populations, tumor microenvironment and systemic determinants of cancer development like the immune system and metabolism. A better appreciation of these differences between the two sexes could be of substantial value for cancer prevention as well as treatment.
    [Show full text]
  • Propranolol-Mediated Attenuation of MMP-9 Excretion in Infants with Hemangiomas
    Supplementary Online Content Thaivalappil S, Bauman N, Saieg A, Movius E, Brown KJ, Preciado D. Propranolol-mediated attenuation of MMP-9 excretion in infants with hemangiomas. JAMA Otolaryngol Head Neck Surg. doi:10.1001/jamaoto.2013.4773 eTable. List of All of the Proteins Identified by Proteomics This supplementary material has been provided by the authors to give readers additional information about their work. © 2013 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 10/01/2021 eTable. List of All of the Proteins Identified by Proteomics Protein Name Prop 12 mo/4 Pred 12 mo/4 Δ Prop to Pred mo mo Myeloperoxidase OS=Homo sapiens GN=MPO 26.00 143.00 ‐117.00 Lactotransferrin OS=Homo sapiens GN=LTF 114.00 205.50 ‐91.50 Matrix metalloproteinase‐9 OS=Homo sapiens GN=MMP9 5.00 36.00 ‐31.00 Neutrophil elastase OS=Homo sapiens GN=ELANE 24.00 48.00 ‐24.00 Bleomycin hydrolase OS=Homo sapiens GN=BLMH 3.00 25.00 ‐22.00 CAP7_HUMAN Azurocidin OS=Homo sapiens GN=AZU1 PE=1 SV=3 4.00 26.00 ‐22.00 S10A8_HUMAN Protein S100‐A8 OS=Homo sapiens GN=S100A8 PE=1 14.67 30.50 ‐15.83 SV=1 IL1F9_HUMAN Interleukin‐1 family member 9 OS=Homo sapiens 1.00 15.00 ‐14.00 GN=IL1F9 PE=1 SV=1 MUC5B_HUMAN Mucin‐5B OS=Homo sapiens GN=MUC5B PE=1 SV=3 2.00 14.00 ‐12.00 MUC4_HUMAN Mucin‐4 OS=Homo sapiens GN=MUC4 PE=1 SV=3 1.00 12.00 ‐11.00 HRG_HUMAN Histidine‐rich glycoprotein OS=Homo sapiens GN=HRG 1.00 12.00 ‐11.00 PE=1 SV=1 TKT_HUMAN Transketolase OS=Homo sapiens GN=TKT PE=1 SV=3 17.00 28.00 ‐11.00 CATG_HUMAN Cathepsin G OS=Homo
    [Show full text]
  • Human Autologous Ipsc–Derived Dopaminergic Progenitors Restore Motor Function in Parkinson’S Disease Models
    Human autologous iPSC–derived dopaminergic progenitors restore motor function in Parkinson’s disease models Bin Song, … , Jeffrey S. Schweitzer, Kwang-Soo Kim J Clin Invest. 2020;130(2):904-920. https://doi.org/10.1172/JCI130767. Research Article Neuroscience Stem cells Graphical abstract Find the latest version: https://jci.me/130767/pdf RESEARCH ARTICLE The Journal of Clinical Investigation Human autologous iPSC–derived dopaminergic progenitors restore motor function in Parkinson’s disease models Bin Song,1,2 Young Cha,1,2 Sanghyeok Ko,1,2 Jeha Jeon,1,2 Nayeon Lee,1,2 Hyemyung Seo,1,2,3 Kyung-Joon Park,1 In-Hee Lee,4,5 Claudia Lopes,1,2 Melissa Feitosa,1,2 María José Luna,1,2 Jin Hyuk Jung,1,2 Jisun Kim,1,2,3 Dabin Hwang,1,2 Bruce M. Cohen,1 Martin H. Teicher,1 Pierre Leblanc,1,2 Bob S. Carter,6 Jeffrey H. Kordower,7 Vadim Y. Bolshakov,1 Sek Won Kong,4,5 Jeffrey S. Schweitzer,6 and Kwang-Soo Kim1,2 1Department of Psychiatry and 2Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA. 3Department of Molecular and Life Sciences, Hanyang University, Ansan, Korea. 4Department of Pediatrics, 5Computational Health Informatics Program, Boston Children’s Hospital, and 6Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. 7Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA. Parkinson’s disease (PD) is a neurodegenerative disorder associated with loss of striatal dopamine, secondary to degeneration of midbrain dopamine (mDA) neurons in the substantia nigra, rendering cell transplantation a promising therapeutic strategy.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Cell Reprogramming Technologies for Treatment And
    CELL REPROGRAMMING TECHNOLOGIES FOR TREATMENT AND UNDERSTANDING OF GENETIC DISORDERS OF MYELIN by ANGELA MARIE LAGER Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy Thesis advisor: Paul J Tesar, PhD Department of Genetics and Genome Sciences CASE WESTERN RESERVE UNIVERSITY May 2015 CASE WESTERN RESERVE UNIVERSITY SCHOOL OF GRADUATE STUDIES We hereby approve the thesis/dissertation of Angela Marie Lager Candidate for the Doctor of Philosophy degree*. (signed) Ronald A Conlon, PhD (Committee Chair) Paul J Tesar, PhD (Advisor) Craig A Hodges, PhD Warren J Alilain, PhD (date) 31 March 2015 *We also certify that written approval has been obtained from any proprietary material contained therein. TABLE OF CONTENTS Table of Contents……………………………………………………………………….1 List of Figures……………………………………………………………………………4 Acknowledgements……………………………………………………………………..7 Abstract…………………………………………………………………………………..8 Chapter 1: Introduction and Background………………………………………..11 1.1 Overview of mammalian oligodendrocyte development in the spinal cord and myelination of the central nervous system…………………..11 1.1.1 Introduction……………………………………………………..11 1.1.2 The establishment of the neuroectoderm and ventral formation of the neural tube…………………………………..12 1.1.3 Ventral patterning of the neural tube and specification of the pMN domain in the spinal cord……………………………….15 1.1.4 Oligodendrocyte progenitor cell production through the process of gliogenesis ………………………………………..16 1.1.5 Oligodendrocyte progenitor cell to oligodendrocyte differentiation…………………………………………………...22
    [Show full text]
  • Regulation of Adult Neurogenesis in Mammalian Brain
    International Journal of Molecular Sciences Review Regulation of Adult Neurogenesis in Mammalian Brain 1,2, 3, 3,4 Maria Victoria Niklison-Chirou y, Massimiliano Agostini y, Ivano Amelio and Gerry Melino 3,* 1 Centre for Therapeutic Innovation (CTI-Bath), Department of Pharmacy & Pharmacology, University of Bath, Bath BA2 7AY, UK; [email protected] 2 Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK 3 Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; [email protected] (M.A.); [email protected] (I.A.) 4 School of Life Sciences, University of Nottingham, Nottingham NG7 2HU, UK * Correspondence: [email protected] These authors contributed equally to this work. y Received: 18 May 2020; Accepted: 7 July 2020; Published: 9 July 2020 Abstract: Adult neurogenesis is a multistage process by which neurons are generated and integrated into existing neuronal circuits. In the adult brain, neurogenesis is mainly localized in two specialized niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) adjacent to the lateral ventricles. Neurogenesis plays a fundamental role in postnatal brain, where it is required for neuronal plasticity. Moreover, perturbation of adult neurogenesis contributes to several human diseases, including cognitive impairment and neurodegenerative diseases. The interplay between extrinsic and intrinsic factors is fundamental in regulating neurogenesis. Over the past decades, several studies on intrinsic pathways, including transcription factors, have highlighted their fundamental role in regulating every stage of neurogenesis. However, it is likely that transcriptional regulation is part of a more sophisticated regulatory network, which includes epigenetic modifications, non-coding RNAs and metabolic pathways.
    [Show full text]
  • Abbit Monoclonal Igg Antibody Against Human, Mouse SOX1
    Anti-Human SOX1 Antibody, Clone EPR4766 Antibodies Rabbit monoclonal IgG antibody against human, mouse SOX1 Catalog #60095 0.1 mL Product Description The EPR4766 antibody reacts with SOX1, also known as SRY (sex determining region Y)-box 1, an ~40 kDa nuclear transcription factor belonging to the SRY-related HMG-box family. SOX1 is involved in regulating development of the central nervous system and is an early marker for neuroectodermal differentiation in the embryo. Its expression is down-regulated during differentiation in many neuronal subtypes. SOX1 is also a useful marker for identifying embryonic and somatic neuronal progenitor cells (NPCs), in concert with other markers such as SOX2 and SOX9. SOX1 is thought to promote self-renewal in NPCs, whereas it functions in other cell types to promote differentiation. There is evidence that SOX1 plays a role in tumor suppression by binding to ß-catenin and attenuating the WNT signaling pathway, and in mice it plays an essential role in lens development by regulating expression of γ-crystallins. The EPR4766 antibody does not cross-react with rat SOX1. Target Antigen Name: SOX1 Alternative Names: Sox-1, SRY (sex determining region Y)-box 1, SRY-related HMG-box Gene ID: 6656 Species Reactivity: Human, Mouse Host Species: Rabbit Clonality: Monoclonal Clone: EPR4766 Isotype: IgG Immunogen: Synthetic peptide corresponding to the partial amino acid sequence of human SOX1 Conjugate: Unconjugated Applications Verified: ICC, IF, IHC, WB Reported: ICC, IF, IHC, WB Special Applications: This antibody clone has been verified for labeling neural stem and progenitor cells grown with STEMdiff™ Neural Induction Medium (Catalog #05835), STEMdiff™ Neural Progenitor Medium (Catalog #05833), NeuroCult™ NS-A Proliferation Kit (Human; Catalog #05751) and NeuroCult™ Proliferation Kit (Mouse; Catalog #05702).
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2003/0082511 A1 Brown Et Al
    US 20030082511A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2003/0082511 A1 Brown et al. (43) Pub. Date: May 1, 2003 (54) IDENTIFICATION OF MODULATORY Publication Classification MOLECULES USING INDUCIBLE PROMOTERS (51) Int. Cl." ............................... C12O 1/00; C12O 1/68 (52) U.S. Cl. ..................................................... 435/4; 435/6 (76) Inventors: Steven J. Brown, San Diego, CA (US); Damien J. Dunnington, San Diego, CA (US); Imran Clark, San Diego, CA (57) ABSTRACT (US) Correspondence Address: Methods for identifying an ion channel modulator, a target David B. Waller & Associates membrane receptor modulator molecule, and other modula 5677 Oberlin Drive tory molecules are disclosed, as well as cells and vectors for Suit 214 use in those methods. A polynucleotide encoding target is San Diego, CA 92121 (US) provided in a cell under control of an inducible promoter, and candidate modulatory molecules are contacted with the (21) Appl. No.: 09/965,201 cell after induction of the promoter to ascertain whether a change in a measurable physiological parameter occurs as a (22) Filed: Sep. 25, 2001 result of the candidate modulatory molecule. Patent Application Publication May 1, 2003 Sheet 1 of 8 US 2003/0082511 A1 KCNC1 cDNA F.G. 1 Patent Application Publication May 1, 2003 Sheet 2 of 8 US 2003/0082511 A1 49 - -9 G C EH H EH N t R M h so as se W M M MP N FIG.2 Patent Application Publication May 1, 2003 Sheet 3 of 8 US 2003/0082511 A1 FG. 3 Patent Application Publication May 1, 2003 Sheet 4 of 8 US 2003/0082511 A1 KCNC1 ITREXCHO KC 150 mM KC 2000000 so 100 mM induced Uninduced Steady state O 100 200 300 400 500 600 700 Time (seconds) FIG.
    [Show full text]